Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 49.554
Filter
1.
Phytomedicine ; 134: 155994, 2024 Nov.
Article in English | MEDLINE | ID: mdl-39243751

ABSTRACT

BACKGROUND: Salvianolic Acid B (SalB) has been proven to delay the progression of atherosclerosis. The therapeutic mechanisms of this compound are unclear. A novel class of short non-coding RNAs, pre-transfer RNA and mature transfer RNA (tsncRNAs) may regulate gene expression. TsncRNAs-sequencing revealed novel therapeutic targets for SalB. This is the first study focusing on tsncRNAs to treat atherosclerosis using SalB. PURPOSE: To explore the potential mechanism of SalB treating atherosclerosis through tsncRNAs. METHODS: Five groups of mice were created at random: control group (CON), atherosclerosis model group (MOD), SalB with high dose-treated group (SABH), SalB with low dose-treated group (SABL), and Simvastatin-treated group (ST). Aortic sinus plaque, body weight and inflammatory cytokines were evaluated. The Illumina NextSeq equipment was used to do expression profiling of tsncRNAs from serum. The targets of tsncRNAs were then predicted using tRNAscan and TargetScan. The KEGG pathway and GO analysis were utilized to forecast the bioinformatics analysis. Potential tsncRNAs and associated mRNAs were validated using quantitative real-time PCR. RESULTS: tRF-Glu-CTC-014 and tRF-Gly-GCC-074 were markedly increased by SalB with high dose treatment and validated with quantitative real-time PCR. Two mRNAs SRF and Arrb related to tRF-Glu-CTC-014 changed consistently. GO analysis revealed that the altered target genes of the selected tsncRNAs were most enriched in protein binding and cellular process. Moreover, KEGG pathway analysis demonstrated that altered target genes of tsncRNAs were most enriched in MAPK signaling pathway. CONCLUSION: SalB can promote the expression of tRF-Glu-CTC-014 to treat atherosclerosis.


Subject(s)
Atherosclerosis , Benzofurans , Mice, Inbred C57BL , Animals , Atherosclerosis/drug therapy , Benzofurans/pharmacology , Male , Mice , Disease Models, Animal , Cytokines/metabolism , Cytokines/blood , Simvastatin/pharmacology , Depsides
2.
Atherosclerosis ; 397: 118567, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39243663

ABSTRACT

BACKGROUND AND AIMS: Mast cell-derived heparin proteoglycans (HEP-PG) can be mimicked by bioconjugates carrying antithrombotic and anti-inflammatory properties. The dual antiplatelet and anticoagulant (APAC) construct administered, either locally or intravenously (i.v.), targets activated endothelium, its adhesion molecules, and subendothelial matrix proteins, all relevant to atherogenesis. We hypothesized that APAC influences cellular interactions in atherosclerotic lesion development and studied APAC treatment during the initiation and progression of experimental atherosclerosis. METHODS: Male western-type diet-fed Apoe-/- mice were equipped with perivascular carotid artery collars to induce local atherosclerosis. In this model, mRNA expression of adhesion molecules including ICAM-1, VCAM-1, P-Selectin, and Platelet Factor 4 (PF4) are upregulated upon lesion development. From day 1 (prevention) or from 2.5 weeks after lesion initiation (treatment), mice were administered 0.2 mg/kg APAC i.v. or control vehicle three times weekly for 2.5 weeks. At week 5 after collar placement, mice were sacrificed, and lesion morphology was microscopically assessed. RESULTS: APAC treatment did not affect body weight or plasma total cholesterol levels during the experiments. In the prevention setting, APAC reduced carotid artery plaque size and volume by over 50 %, aligning with decreased plaque macrophage area and collagen content. During the treatment setting, APAC reduced macrophage accumulation and necrotic core content, and improved markers of plaque stability. CONCLUSIONS: APAC effectively reduced early atherosclerotic lesion development and improved markers of plaque inflammation in advanced atherosclerosis. Thus, APAC may have potential to alleviate the progression of atherosclerosis.


Subject(s)
Anticoagulants , Carotid Artery Diseases , Disease Models, Animal , Mice, Knockout, ApoE , Plaque, Atherosclerotic , Platelet Aggregation Inhibitors , Animals , Male , Carotid Artery Diseases/prevention & control , Carotid Artery Diseases/pathology , Carotid Artery Diseases/metabolism , Carotid Artery Diseases/drug therapy , Anticoagulants/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Mice , Proteoglycans , Carotid Arteries/pathology , Carotid Arteries/drug effects , Mice, Inbred C57BL , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Atherosclerosis/metabolism , Apolipoproteins E/genetics , Platelet Factor 4 , Vascular Cell Adhesion Molecule-1/metabolism , Heparin/analogs & derivatives
3.
Atherosclerosis ; 397: 118582, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39260002

ABSTRACT

BACKGROUND AND AIMS: Lysyl oxidase (LOX) catalyzes the crosslinking of collagen and elastin to maintain tensile strength and structural integrity of the vasculature. Excessive LOX activity increases vascular stiffness and the severity of occlusive diseases. Herein, we investigated the mechanisms by which LOX controls atherogenesis and osteogenic differentiation of vascular smooth muscle cells (SMC) in hyperlipidemic mice. METHODS: Gene inactivation of Lox in SMC was achieved in conditional knockout mice after tamoxifen injections. Atherosclerosis burden and vascular calcification were assessed in hyperlipidemic conditional [Loxf/fMyh11-CreERT2ApoE-/-] and sibling control mice [Loxwt/wtMyh11-CreERT2ApoE-/-]. Mechanistic studies were performed with primary aortic SMC from Lox mutant and wild type mice. RESULTS: Inactivation of Lox in SMCs decreased > 70 % its RNA expression and protein level in the aortic wall and significantly reduced LOX activity without compromising vascular structure and function. Moreover, LOX deficiency protected mice against atherosclerotic burden (13 ± 2 versus 23 ± 1 %, p < 0.01) and plaque calcification (5 ± 0.4 versus 11.8 ± 3 %, p < 0.05) compared to sibling controls. Interestingly, gene inactivation of Lox in SMCs preserved the contractile phenotype of vascular SMC under hyperlipidemic conditions as demonstrated by single-cell RNA sequencing and immunofluorescence. Mechanistically, the absence of LOX in SMC prevented excessive collagen crosslinking and the subsequent activation of the pro-osteogenic FAK/ß-catenin signaling axis. CONCLUSIONS: Lox inactivation in SMC protects mice against atherosclerosis and plaque calcification by reducing SMC modulation and FAK/ß-catenin signaling.


Subject(s)
Atherosclerosis , Disease Models, Animal , Hyperlipidemias , Mice, Knockout , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Plaque, Atherosclerotic , Protein-Lysine 6-Oxidase , Vascular Calcification , Animals , Protein-Lysine 6-Oxidase/metabolism , Protein-Lysine 6-Oxidase/genetics , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/pathology , Myocytes, Smooth Muscle/metabolism , Atherosclerosis/genetics , Atherosclerosis/enzymology , Atherosclerosis/pathology , Atherosclerosis/metabolism , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/metabolism , Vascular Calcification/genetics , Vascular Calcification/pathology , Vascular Calcification/enzymology , Vascular Calcification/prevention & control , Vascular Calcification/metabolism , Hyperlipidemias/genetics , Hyperlipidemias/enzymology , Hyperlipidemias/complications , Hyperlipidemias/metabolism , Mice , Osteogenesis , Cells, Cultured , Aortic Diseases/pathology , Aortic Diseases/genetics , Aortic Diseases/enzymology , Aortic Diseases/prevention & control , Aortic Diseases/metabolism , Aorta/pathology , Aorta/enzymology , Aorta/metabolism , Male , Mice, Inbred C57BL , beta Catenin/metabolism , Signal Transduction , Extracellular Matrix Proteins
4.
Atherosclerosis ; 397: 118570, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39276419

ABSTRACT

BACKGROUND AND AIMS: CCN4/WISP-1 regulates various cell behaviours that contribute to atherosclerosis progression, including cell adhesion, migration, proliferation and survival. We therefore hypothesised that CCN4 regulates the development and progression of atherosclerotic plaques. METHODS: We used a high fat fed ApoE-/- mouse model to study atherosclerotic plaque progression in the brachiocephalic artery and aortic root. In protocol 1, male ApoE-/- mice with established plaques were given a CCN4 helper-dependent adenovirus to see the effect of treatment with CCN4, while in protocol 2 male CCN4-/-ApoE-/- were compared to CCN4+/+ApoE-/- mice to assess the effect of CCN4 deletion on plaque progression. RESULTS: CCN4 overexpression resulted in reduced occlusion of the brachiocephalic artery with less apoptosis, fewer macrophages, and attenuated lipid core size. The amount of plaque found on the aortic root was also reduced. CCN4 deficiency resulted in increased apoptosis and occlusion of the brachiocephalic artery as well as increased plaque in the aortic root. Additionally, in vitro cells from CCN4-/-ApoE-/- mice had higher apoptotic levels. CCN4 deficiency did not significantly affect blood cholesterol levels or circulating myeloid cell populations. CONCLUSIONS: We conclude that in an atherosclerosis model the most important action of CCN4 is the effect on cell apoptosis. CCN4 provides pro-survival signals and leads to reduced cell death, lower macrophage number, smaller lipid core size and reduced atherosclerotic plaque burden. As such, the pro-survival effect of CCN4 is worthy of further investigation, in a bid to find a therapeutic for atherosclerosis.


Subject(s)
Apoptosis , Atherosclerosis , Disease Models, Animal , Mice, Knockout, ApoE , Plaque, Atherosclerotic , Animals , Male , Atherosclerosis/pathology , Atherosclerosis/metabolism , Atherosclerosis/genetics , CCN Intercellular Signaling Proteins/metabolism , CCN Intercellular Signaling Proteins/genetics , Mice, Inbred C57BL , Brachiocephalic Trunk/pathology , Brachiocephalic Trunk/metabolism , Mice , Macrophages/metabolism , Disease Progression , Diet, High-Fat , Apolipoproteins E/deficiency , Apolipoproteins E/genetics , Aorta/pathology , Aorta/metabolism
5.
Turk Kardiyol Dern Ars ; 52(6): 384-389, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39225648

ABSTRACT

OBJECTIVE: This study aims to evaluate the role of elevated lipoprotein (a) [Lp(a)] levels as a potential contributor to residual risk in individuals with atherosclerotic cardiovascular disease (ASCVD). Considering that approximately 90% of Lp(a) levels are genetically determined and can vary regionally, we assessed Lp(a) levels in a cohort of ASCVD patients from the Turkish population, where data is currently limited. METHODS: We conducted a retrospective analysis of data and Lp(a) measurements collected from individuals diagnosed with ASCVD at a single center. RESULTS: The analysis included Lp(a) levels of 1193 consecutive individuals. The mean Lp(a) level was 28.2 mg/dL, with a median of 16 mg/dL and an interquartile range (IQR) from the 25th to the 75th percentile, 7 mg/dL to 39 mg/dL. The highest recorded Lp(a) level was 326 mg/dL. Among the cases, 18.7% exhibited Lp(a) levels ≥ 50 mg/dL, 10.8% had levels ≥ 70 mg/dL, and 5.8% had levels ≥ 90 mg/dL. The mean levels of low-density lipoprotein cholesterol (LDL-C) and total cholesterol (TC) were 132 ± 47 mg/dL and 212 ± 54 mg/dL, respectively. Lp(a) levels were significantly higher in females compared to males. Furthermore, the proportion of females with Lp(a) levels ≥ 90 mg/dL was higher than in males (11.4% vs. 1.4%; P < 0.01). Additionally, a modest but significant correlation was observed between Lp(a) levels and TC (r = 0.075, P = 0.01) as well as LDL-C (r = 0.106, P < 0.01). CONCLUSION: This study revealed that Lp(a) concentrations were higher in women and statin users among ASCVD patients and identified a weak but significant correlation between Lp(a) levels and both TC and LDL-C.


Subject(s)
Atherosclerosis , Lipoprotein(a) , Humans , Male , Female , Lipoprotein(a)/blood , Retrospective Studies , Turkey/epidemiology , Middle Aged , Atherosclerosis/blood , Aged , Adult
6.
BMC Infect Dis ; 24(1): 932, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39251924

ABSTRACT

BACKGROUND: Cardiovascular disease is a major cause of morbidity in an aging HIV population. However, risk estimation with the most frequent equations usually classifies HIV patients as having a low or moderate risk. Several studies have described a very high prevalence of subclinical atherosclerosis in a middle-aged, non-HIV population. There is insufficient body of knowledge to understand if this is the case in people living with HIV (PLWH). We aim to calculate the proportion of patients with subclinical atherosclerosis in a single site cohort of HIV-infected subjects. METHODS: We have analyzed chronically HIV infected adults (≥ 18 years) who were on active follow-up in an HIV unit specialized in the care of cardiovascular health. The most recent clinical visit and vascular ultrasonography were used to assess the objectives of our research. Our primary objective was to describe the proportion of participants with subclinical atherosclerosis (focal protrusion into the lumen > 0.5 mm or > 50% of the surrounding IMT or a diffuse thickness > 1.5 mm) in a single site cohort of PLWH. Carotid and iliofemoral territories were evaluated. As a secondary objective we have run a multivariate analysis to determine which HIV and non-HIV factors might be related with the presence of atherosclerotic plaques. Findings We included a total of 463 participants between November 2017 to October 2019. Subjects were predominantly male (84.2%) with a mean age of 48.8 years (SD 10.7). Hypercholesterolemia (36%) was the most prevalent comorbidity followed by Hypertension (18%) and Hypertriglyceridemia (16%). Mean duration of HIV infection is 12.3 years. Overall, participants had been receiving cART for a median of 9.5 years. Subclinical atherosclerosis was found in 197 subjects (42.5%; CI 95% [38.0-47.2]). The disease was found more frequently in the femoral arteries (37.8%) than in the carotid vascular bed (18.6%). Despite some HIV factors correlated with the presence of plaques in a univariate analysis (e.g., time with HIV-1 RNA > 50 copies/mL or time from HIV diagnosis), the only two explanatory factors that remained associated with the presence of atherosclerotic plaques in the multivariate analysis were smoking (OR 5.47, 95% CI 3.36 - 8.90) and age (OR 1.13, 95%CI 1.10 - 1.16). Interpretation We have found a very high prevalence of subclinical atherosclerosis among our cohort of PLWH. Despite having analyzed several HIV factors, age and smoking have been found to be the only factors associated with the development of atherosclerotic plaques.


Subject(s)
Atherosclerosis , Femoral Artery , HIV Infections , Humans , Male , HIV Infections/complications , HIV Infections/epidemiology , Middle Aged , Female , Atherosclerosis/epidemiology , Adult , Risk Factors , Femoral Artery/diagnostic imaging , Femoral Artery/pathology , Prevalence , Carotid Arteries/diagnostic imaging , Carotid Arteries/pathology , Cohort Studies
7.
Cardiovasc Diabetol ; 23(1): 331, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39252021

ABSTRACT

BACKGROUND: Visceral adipose tissue in individuals with obesity is an independent cardiovascular risk indicator. However, it remains unclear whether adipose tissue influences common cardiovascular diseases, such as atherosclerosis, through its secreted exosomes. METHODS: The exosomes secreted by adipose tissue from diet-induced obesity mice were isolated to examine their impact on the progression of atherosclerosis and the associated mechanism. Endothelial apoptosis and the proliferation and migration of vascular smooth muscle cells (VSMCs) within the atherosclerotic plaque were evaluated. Statistical significance was analyzed using GraphPad Prism 9.0 with appropriate statistical tests. RESULTS: We demonstrate that adipose tissue-derived exosomes (AT-EX) exacerbate atherosclerosis progression by promoting endothelial apoptosis, proliferation, and migration of VSMCs within the plaque in vivo. MicroRNA-132/212 (miR-132/212) was detected within AT-EX cargo. Mechanistically, miR-132/212-enriched AT-EX exacerbates palmitate acid-induced endothelial apoptosis via targeting G protein subunit alpha 12 and enhances platelet-derived growth factor type BB-induced VSMC proliferation and migration by targeting phosphatase and tensin homolog in vitro. Importantly, melatonin decreases exosomal miR-132/212 levels, thereby mitigating the pro-atherosclerotic impact of AT-EX. CONCLUSION: These data uncover the pathological mechanism by which adipose tissue-derived exosomes regulate the progression of atherosclerosis and identify miR-132/212 as potential diagnostic and therapeutic targets for atherosclerosis.


Subject(s)
Apoptosis , Atherosclerosis , Cell Movement , Cell Proliferation , Disease Models, Animal , Disease Progression , Exosomes , Mice, Inbred C57BL , MicroRNAs , Muscle, Smooth, Vascular , Myocytes, Smooth Muscle , Plaque, Atherosclerotic , Animals , MicroRNAs/metabolism , MicroRNAs/genetics , Exosomes/metabolism , Exosomes/pathology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Atherosclerosis/genetics , Cell Proliferation/drug effects , Apoptosis/drug effects , Myocytes, Smooth Muscle/metabolism , Myocytes, Smooth Muscle/pathology , Myocytes, Smooth Muscle/drug effects , Cell Movement/drug effects , Muscle, Smooth, Vascular/pathology , Muscle, Smooth, Vascular/metabolism , Muscle, Smooth, Vascular/drug effects , Male , Signal Transduction , Cells, Cultured , Obesity/metabolism , Obesity/pathology , Mice, Knockout, ApoE , Endothelial Cells/metabolism , Endothelial Cells/pathology , Endothelial Cells/drug effects , Aortic Diseases/pathology , Aortic Diseases/metabolism , Aortic Diseases/genetics , Becaplermin/pharmacology , Becaplermin/metabolism , Intra-Abdominal Fat/metabolism , Intra-Abdominal Fat/pathology , Mice , Humans
8.
J Am Coll Cardiol ; 84(14): 1328-1338, 2024 Oct 01.
Article in English | MEDLINE | ID: mdl-39322327

ABSTRACT

BACKGROUND: Triglyceride-rich lipoproteins and remnants (TRL/remnants) have a causal, but not yet quantified, relationship with coronary heart disease (CHD): myocardial infarction plus revascularization. OBJECTIVES: The authors sought to estimate TRL/remnant per-particle atherogenicity, investigate causal relationships with inflammation, and determine whether differences in the atherogenicity of TRL/remnants and low-density lipoprotein (LDL) impact the causal association of non-high-density lipoprotein cholesterol (non-HDL-C) with CHD. METHODS: Single nucleotide polymorphisms (SNPs) (N = 1,357) identified by genome-wide association in the UK Biobank were ranked into 10 clusters according to the effect on TRL/remnant-C vs LDL-C. Mendelian randomization analysis was used to estimate for each SNP cluster CHD ORs per 10 mg/dL apolipoprotein B (apoB) and per 0.33 mmol/L non-HDL-cholesterol, and to evaluate association of TRL/remnants with biomarkers of systemic inflammation. RESULTS: SNPs in cluster 1 predominantly affected LDL-C, whereas SNPs in cluster 10 predominantly affected TRL/remnant-C. CHD risk per genetically predicted increase in apoB and in non-HDL-C rose across clusters. ORs per 10 mg/dL higher apoB was 1.15 (95% CI: 1.11-1.19) in cluster 1 vs 1.70 (95% CI: 1.52-1.90) in cluster 10. Comparing ORs between these TRL/remnant-predominant and LDL-predominant clusters, we estimated that TRL/remnants were at least 3.9 (95% CI: 2.8-5.4) times more atherogenic than LDL on a per-particle basis. For non-HDL-C, CHD ORs per 0.33 mmol/L rose from 1.15 (95% CI: 1.11-1.19) for cluster 1 to 1.40 (95% CI: 1.30-1.50) for cluster 10. TRL/remnants exhibited causal relationships with inflammation, but this did not explain their greater atherogenicity. CONCLUSIONS: TRL/remnants are about 4 times more atherogenic than LDL. Variation in the causal association of non-HDL-C with CHD indicates that adjustment for percentage TRL/remnant-C may be needed for accurate risk prediction.


Subject(s)
Inflammation , Polymorphism, Single Nucleotide , Triglycerides , Humans , Triglycerides/blood , Inflammation/blood , Inflammation/genetics , Male , Risk Assessment/methods , Female , Middle Aged , Atherosclerosis/blood , Atherosclerosis/epidemiology , Atherosclerosis/genetics , Lipoproteins/blood , Cholesterol/blood , Coronary Disease/blood , Coronary Disease/genetics , Coronary Disease/epidemiology , Genome-Wide Association Study , Mendelian Randomization Analysis , Aged , Cholesterol, LDL/blood , Biomarkers/blood , Cholesterol, HDL/blood , United Kingdom/epidemiology
10.
Ren Fail ; 46(2): 2407885, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39324542

ABSTRACT

BACKGROUND: Patients receiving maintenance hemodialysis (MHD) frequently encounter a drop in blood pressure during dialysis, known as intradialytic hypotension (IDH). The AIP is associated with diseases such as diabetes and cardiovascular events. It remains unclear whether the AIP is associated with IDH. The present study aimed to explore the association between AIP and IDH in MHD patients. METHODS: In this multi-center cross-sectional study, we included 1946 adult hemodialysis patients from twenty dialysis centers. Patients were divided into four groups based on the AIP quartiles. Linear regression and multiple logistic regression models were used to analyze the relationship between AIP and IDH. Subgroup analyses were further conducted to assess the robustness of the association between the AIP and IDH. RESULTS: After adjusting for potential confounding variables, each 1-unit increase in AIP was associated with a 21% increase in the odds of IDH. The odds ratios (ORs) of IDH increased gradually with higher quartiles of AIP compared with the Q1 reference group (Q2: OR, 1.41, 95% CI: 0.91-2.18; Q3: OR, 1.63, 95% CI: 1.07-2.49; Q4: OR, 1.57, 95% CI: 1.01-2.42). No interaction was observed in the subgroup analysis stratified by age, sex, history of diabetes, heart failure, and myocardial infarction. CONCLUSION: Elevated AIP levels are associated with a heightened risk of IDH in MHD patients.


Subject(s)
Hypotension , Renal Dialysis , Humans , Cross-Sectional Studies , Female , Male , Hypotension/etiology , Renal Dialysis/adverse effects , Middle Aged , Aged , Risk Factors , Kidney Failure, Chronic/therapy , Kidney Failure, Chronic/blood , Kidney Failure, Chronic/complications , Logistic Models , Atherosclerosis/etiology , Atherosclerosis/blood , Adult , Blood Pressure
12.
Nat Metab ; 6(9): 1736-1755, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39242914

ABSTRACT

Macrophage efferocytosis prevents apoptotic cell (AC) accumulation and triggers inflammation-resolution pathways. The mechanisms linking efferocytosis to resolution often involve changes in macrophage metabolism, but many gaps remain in our understanding of these processes. We now report that efferocytosis triggers an indoleamine 2,3-dioxygenase-1 (IDO1)-dependent tryptophan (Trp) metabolism pathway that promotes several key resolution processes, including the induction of pro-resolving proteins, such interleukin-10, and further enhancement of efferocytosis. The process begins with upregulation of Trp transport and metabolism, and it involves subsequent activation of the aryl hydrocarbon receptor (AhR) by the Trp metabolite kynurenine (Kyn). Through these mechanisms, macrophage IDO1 and AhR contribute to a proper resolution response in several different mouse models of efferocytosis-dependent tissue repair, notably during atherosclerosis regression induced by plasma low-density lipoprotein (LDL) lowering. These findings reveal an integrated metabolism programme in macrophages that links efferocytosis to resolution, with possible therapeutic implications for non-resolving chronic inflammatory diseases, notably atherosclerosis.


Subject(s)
Indoleamine-Pyrrole 2,3,-Dioxygenase , Macrophages , Phagocytosis , Receptors, Aryl Hydrocarbon , Tryptophan , Tryptophan/metabolism , Animals , Macrophages/metabolism , Mice , Receptors, Aryl Hydrocarbon/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Kynurenine/metabolism , Inflammation/metabolism , Apoptosis , Atherosclerosis/metabolism , Efferocytosis
13.
Expert Rev Cardiovasc Ther ; 22(8): 353-366, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39258965

ABSTRACT

INTRODUCTION: Lipoprotein(a) [Lp(a)] is linked to higher risks of atherosclerotic cardiovascular disease (ASCVD). Current guideline recommendations are quite liberal on measuring Lp(a) (Class IIa, Level C), and may lead to underuse among (interventional) cardiologists. AREAS COVERED: This case-based narrative review outlines four clinical cases of patients with elevated Lp(a) to illustrate its pathophysiological impact on coronary artery disease (CAD). The expert consensus statements from the American Heart Association (AHA) and European Atherosclerosis Society (EAS) served as the basis of this review. More recent publications, from 2023 to 2024, were accessed through the MEDLINE online library. EXPERT OPINION: We highlighted the importance of routine Lp(a) measurement in identifying patients at high risk for atherosclerosis, necessitating potent risk mitigation. Measuring Lp(a) helps clinicians identify which patients are at highest residual risk, who require potent pharmacological treatment and special attention during catheter interventions. As noninvasive and advanced intravascular imaging modalities evolve, future catheterization laboratories will integrate advanced imaging, diagnostics, and treatment, facilitating tailored patient care. Knowing Lp(a) levels is crucial in this context. While Lp(a)-lowering drugs are currently investigated in clinical trials, it is of paramount importance to know Lp(a) levels and strive toward aggressive management of other modifiable risk factors in patients with elevated Lp(a) and established symptomatic CAD being diagnosed or treated in catheterization laboratories.


Subject(s)
Coronary Artery Disease , Lipoprotein(a) , Humans , Atherosclerosis/blood , Atherosclerosis/diagnosis , Biomarkers/blood , Coronary Artery Disease/blood , Coronary Artery Disease/diagnosis , Lipoprotein(a)/blood , Practice Guidelines as Topic , Recurrence , Risk Assessment/methods , Risk Assessment/standards
14.
Cochrane Database Syst Rev ; 9: CD014741, 2024 Sep 19.
Article in English | MEDLINE | ID: mdl-39297531

ABSTRACT

BACKGROUND: Atherosclerotic cardiovascular disease (ACVD) is worsened by chronic inflammatory diseases. Interleukin receptor antagonists (IL-RAs) and tumour necrosis factor-alpha (TNF) inhibitors have been studied to see if they can prevent cardiovascular events. OBJECTIVES: The purpose of this study was to assess the clinical benefits and harms of IL-RAs and TNF inhibitors in the primary and secondary prevention of ACVD. SEARCH METHODS: The Cochrane Heart Specialised Register, the Cochrane Central Register of Controlled Trials (CENTRAL), Ovid MEDLINE (including In-Process & Other Non-Indexed Citations), Ovid Embase, EBSCO CINAHL plus, and clinical trial registries for ongoing and unpublished studies were searched in February 2024. The reference lists of relevant studies, reviews, meta-analyses and health technology reports were searched to identify additional studies. No limitations on language, date of publication or study type were set. SELECTION CRITERIA: RCTs that recruited people with and without pre-existing ACVD, comparing IL-RAs or TNF inhibitors versus placebo or usual care, were selected. The primary outcomes considered were all-cause mortality, myocardial infarction, unstable angina, and adverse events. DATA COLLECTION AND ANALYSIS: Two or more review authors, working independently at each step, selected studies, extracted data, assessed the risk of bias and used GRADE to judge the certainty of evidence. MAIN RESULTS: We included 58 RCTs (22,053 participants; 21,308 analysed), comparing medication efficacy with placebo or usual care. Thirty-four trials focused on primary prevention and 24 on secondary prevention. The interventions included IL-1 RAs (anakinra, canakinumab), IL-6 RA (tocilizumab), TNF-inhibitors (etanercept, infliximab) compared with placebo or usual care. The certainty of evidence was low to very low due to biases and imprecision; all trials had a high risk of bias. Primary prevention: IL-1 RAs The evidence is very uncertain about the effects of the intervention on all-cause mortality(RR 0.33, 95% CI 0.01 to 7.58, 1 trial), myocardial infarction (RR 0.71, 95% CI 0.04 to 12.48, I² = 39%, 2 trials), unstable angina (RR 0.24, 95% CI 0.03 to 2.11, I² = 0%, 2 trials), stroke (RR 2.42, 95% CI 0.12 to 50.15; 1 trial), adverse events (RR 0.85, 95% CI 0.59 to 1.22, I² = 54%, 3 trials), or infection (rate ratio 0.84, 95% 0.55 to 1.29, I² = 0%, 4 trials). Evidence is very uncertain about whether anakinra and cankinumab may reduce heart failure (RR 0.21, 95% CI 0.05 to 0.94, I² = 0%, 3 trials). Peripheral vascular disease (PVD) was not reported as an outcome. IL-6 RAs The evidence is very uncertain about the effects of the intervention on all-cause mortality (RR 0.68, 95% CI 0.12 to 3.74, I² = 30%, 3 trials), myocardial infarction (RR 0.27, 95% CI 0.04 to1.68, I² = 0%, 3 trials), heart failure (RR 1.02, 95% CI 0.11 to 9.63, I² = 0%, 2 trials), PVD (RR 2.94, 95% CI 0.12 to 71.47, 1 trial), stroke (RR 0.34, 95% CI 0.01 to 8.14, 1 trial), or any infection (rate ratio 1.10, 95% CI: 0.88 to 1.37, I2 = 18%, 5 trials). Adverse events may increase (RR 1.13, 95% CI 1.04 to 1.23, I² = 33%, 5 trials). No trial assessed unstable angina. TNF inhibitors The evidence is very uncertain about the effects of the intervention on all-cause mortality (RR 1.78, 95% CI 0.63 to 4.99, I² = 10%, 3 trials), myocardial infarction (RR 2.61, 95% CI 0.11 to 62.26, 1 trial), stroke (RR 0.46, 95% CI 0.08 to 2.80, I² = 0%; 3 trials), heart failure (RR 0.85, 95% CI 0.06 to 12.76, 1 trial). Adverse events may increase (RR 1.13, 95% CI 1.01 to 1.25, I² = 51%, 13 trials). No trial assessed unstable angina or PVD. Secondary prevention: IL-1 RAs The evidence is very uncertain about the effects of the intervention on all-cause mortality (RR 0.94, 95% CI 0.84 to 1.06, I² = 0%, 8 trials), unstable angina (RR 0.88, 95% CI 0.65 to 1.19, I² = 0%, 3 trials), PVD (RR 0.85, 95% CI 0.19 to 3.73, I² = 38%, 3 trials), stroke (RR 0.94, 95% CI 0.74 to 1.2, I² = 0%; 7 trials), heart failure (RR 0.91, 95% 0.5 to 1.65, I² = 0%; 7 trials), or adverse events (RR 0.92, 95% CI 0.78 to 1.09, I² = 3%, 4 trials). There may be little to no difference between the groups in myocardial infarction (RR 0.88, 95% CI 0.0.75 to 1.04, I² = 0%, 6 trials). IL6-RAs The evidence is very uncertain about the effects of the intervention on all-cause mortality (RR 1.09, 95% CI 0.61 to 1.96, I² = 0%, 2 trials), myocardial infarction (RR 0.46, 95% CI 0.07 to 3.04, I² = 45%, 3 trials), unstable angina (RR 0.33, 95% CI 0.01 to 8.02, 1 trial), stroke (RR 1.03, 95% CI 0.07 to 16.25, 1 trial), adverse events (RR 0.89, 95% CI 0.76 to 1.05, I² = 0%, 2 trials), or any infection (rate ratio 0.66, 95% CI 0.32 to 1.36, I² = 0%, 4 trials). No trial assessed PVD or heart failure. TNF inhibitors The evidence is very uncertain about the effect of the intervention on all-cause mortality (RR 1.16, 95% CI 0.69 to 1.95, I² = 47%, 5 trials), heart failure (RR 0.92, 95% 0.75 to 1.14, I² = 0%, 4 trials), or adverse events (RR 1.15, 95% CI 0.84 to 1.56, I² = 32%, 2 trials). No trial assessed myocardial infarction, unstable angina, PVD or stroke. Adverse events may be underestimated and benefits inflated due to inadequate reporting. AUTHORS' CONCLUSIONS: This Cochrane review assessed the benefits and harms of using interleukin-receptor antagonists and tumour necrosis factor inhibitors for primary and secondary prevention of atherosclerotic diseases compared with placebo or usual care. However, the evidence for the predetermined outcomes was deemed low or very low certainty, so there is still a need to determine whether these interventions provide clinical benefits or cause harm from this perspective. In summary, the different biases and imprecision in the included studies limit their external validity and represent a limitation to determining the effectiveness of the intervention for both primary and secondary prevention of ACVD.


Subject(s)
Antibodies, Monoclonal, Humanized , Atherosclerosis , Myocardial Infarction , Primary Prevention , Randomized Controlled Trials as Topic , Secondary Prevention , Tumor Necrosis Factor-alpha , Humans , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Primary Prevention/methods , Myocardial Infarction/prevention & control , Myocardial Infarction/mortality , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/adverse effects , Atherosclerosis/prevention & control , Atherosclerosis/mortality , Cause of Death , Angina, Unstable/prevention & control , Angina, Unstable/mortality , Interleukin 1 Receptor Antagonist Protein/therapeutic use , Interleukin 1 Receptor Antagonist Protein/adverse effects , Cardiovascular Diseases/prevention & control , Cardiovascular Diseases/mortality , Bias
15.
Atherosclerosis ; 397: 118556, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39222595

ABSTRACT

BACKGROUND AND AIMS: Atherosclerotic cardiovascular disease complicated by diabetes mellitus (DM) is the leading cause of death in diabetic patients, and it is strongly associated with macrophages and inflammasomes. It has been found that activation of NOD-like receptor thermal protein domain associated protein 3 (NLRP3) inflammasome is closely associated with phosphatidylinositol 4-phosphate (PI4P) on the trans-Golgi. However, how PI4P and NLRP3 regulate macrophage function and its role in diabetic atherosclerotic plaques is unclear. METHODS: The expression of Pi4p and Nlrp3-inflammasome-related proteins in atherosclerosis in apolipoprotein E-deficient (Apoe-/-) and Apoe-/- DM mice was investigated. Then, Pi4p levels were affected by shRNA-Pi4kb or cDNA-Sac1 plasmid to investigate the effects of changes in Pi4p-related metabolic enzymes on macrophage function. Finally, genetically modified macrophages were injected into diabetic Apoe-/- mice to explore the effects on atherosclerosis. RESULTS: DM promoted plaque progression in atherosclerotic mice and increased expression of Pi4p and Nlrp3 in plaques. In addition, impaired macrophage function induced by high glucose was reversed by transfected shRNA-Pi4kb or cDNA-Sac1 plasmid. Furthermore, decreased levels of Pi4p reduced plaque area in diabetic Apoe-/- mice. CONCLUSIONS: Our data suggests that Pi4p/Nlrp3 in macrophages play an important role in the exacerbation of atherosclerosis in diabetic mice. Pi4p-related metabolizing enzymes (PI4KB and SAC1) may be a potential therapeutic strategy for diabetic atherosclerosis, and macrophage therapy is also a potential treatment.


Subject(s)
Atherosclerosis , Diabetes Mellitus, Experimental , Disease Progression , Macrophages , Mice, Knockout, ApoE , NLR Family, Pyrin Domain-Containing 3 Protein , Plaque, Atherosclerotic , Signal Transduction , Animals , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , Macrophages/metabolism , Atherosclerosis/metabolism , Atherosclerosis/genetics , Atherosclerosis/pathology , Diabetes Mellitus, Experimental/metabolism , Mice , Mice, Inbred C57BL , Male , Inflammasomes/metabolism , Apolipoproteins E/genetics , Apolipoproteins E/deficiency
16.
Adv Exp Med Biol ; 1460: 575-594, 2024.
Article in English | MEDLINE | ID: mdl-39287865

ABSTRACT

Lipotoxicity can mediate endothelial dysfunction in obesity. Altered endothelial cell phenotype during the pathobiological course of the lipotoxicity may lead to hemostatic abnormalities, which is a hallmark of several hematological disorders. Impaired hemostasis could also be directly related to numerous metabolic diseases such as hypertension, diabetes, and atherosclerosis. On the other hand, the local hematopoietic bone marrow (BM) renin-angiotensin system (RAS) contributes to the development of atherosclerosis via acting on the lipotoxicity processes. Local BM RAS, principally an autocrine/paracrine/intracrine hematological system, is located at the crossroads of cellular regulation, molecular interactions, and lipotoxicity-mediated vascular endothelial dysfunction. The positive regulatory role of plasma LDL on AT1 receptor-mediated hematopoietic stem cell (HSC) differentiation and the production of pro-atherogenic monocytes have been described. LDL-regulated HSC function may explain in part hypercholesterolemia-induced inflammation as well as the anti-inflammatory and anti-atherosclerotic effects of AT1 receptor blockers. The role of local adipose tissue RAS is directly related to the pathogenesis of metabolic derangements in obesity. There may be a crosstalk between local BM RAS and local adipose tissue RAS at the genomics and transcriptomics levels. This chapter aims to review hematological alterations propagating the pathological influences of lipotoxicity on the vascular endothelium.


Subject(s)
Hematologic Diseases , Obesity , Renin-Angiotensin System , Humans , Obesity/metabolism , Obesity/complications , Hematologic Diseases/metabolism , Hematologic Diseases/pathology , Hematologic Diseases/etiology , Renin-Angiotensin System/physiology , Animals , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Adipose Tissue/metabolism , Adipose Tissue/pathology , Atherosclerosis/metabolism , Atherosclerosis/pathology , Atherosclerosis/etiology
20.
Cells ; 13(18)2024 Sep 21.
Article in English | MEDLINE | ID: mdl-39329770

ABSTRACT

Atherosclerosis is an autoimmune disease characterized by lipid imbalances and chronic inflammation within blood vessels, with limited preventive and treatment options currently available. In this study, a vaccine prepared with COL6A6 peptide (named the Pep_A6 vaccine) was administered to immunize Apoe-/- mice, and the immune mechanism of the Pep_A6 vaccine against atherosclerosis was first investigated. The results of arterial oil red O staining demonstrated that the Pep_A6 vaccine significantly reduced the atherosclerotic plaque area in Apoe-/- mice fed with a high-fat diet for 20 weeks. A flow cytometry analysis revealed that the Pep_A6 vaccine inhibited Th1 cell differentiation and increased the proportion of Treg cells. Furthermore, there was a significant increase in Ly6Clow monocytes observed in the vaccinated group. The ELISA results showed that the Pep_A6 vaccine induced a significant expression of Pep_A6-specific antibody IgG and IgG1 in mouse serum. Additionally, we found that the Pep_A6 vaccine significantly decreased serum LDL-C content and regulated the expression of genes related to liver lipid metabolism. Together, our findings suggest that the Pep_A6 vaccine alleviates atherosclerosis by inducing a positive immune response and regulating lipid metabolism, providing new insights into potential prevention strategies for atherosclerosis as an innovative vaccine.


Subject(s)
Apolipoproteins E , Atherosclerosis , Collagen Type VI , Lipid Metabolism , Vaccines, Subunit , Animals , Atherosclerosis/immunology , Atherosclerosis/prevention & control , Mice , Apolipoproteins E/deficiency , Apolipoproteins E/metabolism , Vaccines, Subunit/immunology , Collagen Type VI/metabolism , Mice, Inbred C57BL , Plaque, Atherosclerotic/immunology , Male , Immunity , Mice, Knockout , Diet, High-Fat , Immunoglobulin G/blood , Immunoglobulin G/immunology , Disease Models, Animal , Th1 Cells/immunology , Protein Subunit Vaccines
SELECTION OF CITATIONS
SEARCH DETAIL